Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Neurobiol Aging ; 135: 60-69, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38185053

ABSTRACT

Alzheimer's disease (AD) is more prevalent in women than men, supposing due to the decline of estrogens in menopause, accompanied by increased gonadotropins such as luteinizing hormone (LH). We and others found that the transcription factor early growth response-1 (EGR1) regulates cholinergic function including the expression of acetylcholinesterase (AChE) and plays a significant role in cognitive decline of AD. Here we investigated in APP/PS1 mice by ovariectomy (OVX) and estradiol (E2) supplementation or inhibition of LH the effect on hippocampus-related cognition and related molecular changes. We found that OVX-associated cognitive impairment was accompanied by increased dorsal hippocampal EGR1 expression, which was rescued by downregulating peripheral LH rather than by supplementing E2. We also found in postmortem AD brains a higher expression of pituitary LH-mRNA and higher EGR1 expression in the posterior hippocampus. Both, in human and mice, there was a significant positive correlation between respectively posterior/dorsal hippocampal EGR1 and peripheral LH expression. We conclude that peripheral increased LH and increased posterior hippocampal EGR1 plays a significant role in AD pathology.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Female , Animals , Humans , Luteinizing Hormone/metabolism , Down-Regulation , Acetylcholinesterase , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Alzheimer Disease/metabolism , Cognition , Ovariectomy , Mice, Transgenic , Disease Models, Animal , Hippocampus/metabolism
2.
Handb Clin Neurol ; 181: 427-443, 2021.
Article in English | MEDLINE | ID: mdl-34238476

ABSTRACT

Gender identity (an individual's perception of being male or female) and sexual orientation (heterosexuality, homosexuality, or bisexuality) are programmed into our brain during early development. During the intrauterine period in the second half of pregnancy, a testosterone surge masculinizes the fetal male brain. If such a testosterone surge does not occur, this will result in a feminine brain. As sexual differentiation of the brain takes place at a much later stage in development than sexual differentiation of the genitals, these two processes can be influenced independently of each other and can result in gender dysphoria. Nature produces a great variability for all aspects of sexual differentiation of the brain. Mechanisms involved in sexual differentiation of the brain include hormones, genetics, epigenetics, endocrine disruptors, immune response, and self-organization. Furthermore, structural and functional differences in the hypothalamus relating to gender dysphoria and sexual orientation are described in this review. All the genetic, postmortem, and in vivo scanning observations support the neurobiological theory about the origin of gender dysphoria, i.e., it is the sizes of brain structures, the neuron numbers, the molecular composition, functions, and connectivity of brain structures that determine our gender identity or sexual orientation. There is no evidence that one's postnatal social environment plays a crucial role in the development of gender identity or sexual orientation.


Subject(s)
Gender Identity , Transsexualism , Female , Humans , Hypothalamus , Male , Pregnancy , Sex Differentiation , Sexual Behavior
3.
Handb Clin Neurol ; 179: 141-156, 2021.
Article in English | MEDLINE | ID: mdl-34225959

ABSTRACT

The quality of postmortem hypothalamus research depends strongly on a thorough clinical investigation and documentation of the patient's disorder and therapies. In addition, a systematic and professional neuropathological investigation of the entire brain of both the cases and the controls is absolutely crucial. In the experience of the Netherlands Brain Bank (NBB), about 20% of the clinical neurological diagnoses, despite being made in first rate clinics, have to be revised or require extra diagnoses after a complete and thorough neuropathologic review by the NBB. The neuropathology examination may reveal for instance that the elderly "controls" already have preclinical neurodegenerative alterations. In postmortem studies, the patient and control groups must be matched for as many as possible of the known confounding factors. This is necessary to make the groups as similar as possible, except for the topic being investigated. Confounding factors are present (i) before, (ii) during, and (iii) after death. They are, respectively: (i) genetic background, systemic diseases, duration and gravity of illness, medicines and addictive compounds used, age, sex, gender identity, sexual orientation, clock- and seasonal time of death, and lateralization; (ii) agonal state, stress of dying; and (iii) postmortem delay, freezing procedures, fixation, and storage time. Agonal state is generally estimated by measuring the pH of the brain. However, there are disorders in which pH is lower as a part of the disease process. Because of the large number of potentially confounding factors that differ according to, for instance, brain area and disease, a brain bank should have a large number of controls at its disposal for appropriate matching. If matching fails for some confounders, the influence of the confounders may be determined by statistical methods, such as analysis of variance or the regression models.


Subject(s)
Gender Identity , Postmortem Changes , Aged , Brain , Female , Humans , Hypothalamus , Male , Netherlands , Neuropathology
4.
Schizophr Bull ; 47(5): 1310-1319, 2021 08 21.
Article in English | MEDLINE | ID: mdl-33974073

ABSTRACT

Hypocretin (also called orexin) regulates various functions, such as sleep-wake rhythms, attention, cognition, and energy balance, which show significant changes in schizophrenia (SCZ). We aimed to identify alterations in the hypocretin system in SCZ patients. We measured plasma hypocretin-1 levels in SCZ patients and healthy controls and found significantly decreased plasma hypocretin-1 levels in SCZ patients, which was mainly due to a significant decrease in female SCZ patients compared with female controls. In addition, we measured postmortem hypothalamic hypocretin-1-immunoreactivity (ir), ventricular cerebrospinal fluid (CSF) hypocretin-1 levels, and hypocretin receptor (Hcrt-R) mRNA expression in the superior frontal gyrus (SFG) in SCZ patients and controls We observed a significant decrease in the amount of hypothalamic hypocretin-1 ir in SCZ patients, which was due to decreased amounts in female but not male patients. Moreover, Hcrt-R2 mRNA in the SFG was decreased in female SCZ patients compared with female controls, while male SCZ patients showed a trend of increased Hcrt-R1 mRNA and Hcrt-R2 mRNA expression compared with male controls. We conclude that central hypocretin neurotransmission is decreased in SCZ patients, especially female patients, and this is reflected in the plasma.


Subject(s)
Hypothalamus/metabolism , Orexin Receptors/metabolism , Orexins/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/metabolism , Adult , Autopsy , Female , Humans , Male , Middle Aged , Orexins/blood , Schizophrenia/blood , Sex Factors
5.
Brain Pathol ; 29(4): 502-512, 2019 07.
Article in English | MEDLINE | ID: mdl-30511454

ABSTRACT

Our previous studies showed that the transcription factor early growth response-1 (EGR1) may play a role in keeping the brain cholinergic function intact in the preclinical stages of Alzheimer's disease (AD). In order to elucidate the mechanisms involved, we first performed data mining on our previous microarray study on postmortem human prefrontal cortex (PFC) for the changes in the expression of EGR1 and acetylcholinesterase (AChE) and the relationship between them during the course of AD. The study contained 49 patients, ranging from non-demented controls (Braak stage 0) to late AD patients (Braak stage VI). We found EGR1-mRNA was high in early AD and decreased in late AD stages, while AChE-mRNA was stable in preclinical AD and slightly decreased in late AD stages. A significant positive correlation was found between the mRNA levels of these two molecules. In addition, we studied the relationship between EGR1 and AChE mRNA levels in the frontal cortex of 3-12-months old triple-transgenic AD (3xTg-AD) mice. EGR1- and AChE-mRNA were lower in 3xTg-AD mice compared with wild-type (WT) mice. A significant positive correlation between these two molecules was present in the entire group and in each age group of either WT or 3xTg-AD mice. Subsequently, AChE expression was determined following up- or down-regulating EGR1 in cell lines and the EGR1 levels were found to regulate AChE at both the mRNA and protein levels. Dual-luciferase assay and electrophoretic mobility shift assay in the EGR1-overexpressing cells were performed to determine the functionally effective binding sites of the EGR1 on the AChE gene promoter. We conclude that the EGR1 can upregulate AChE expression by a direct effect on its gene promoter, which may contribute significantly to the changes in cholinergic function in the course of AD. The 3xTg-AD mouse model only reflects later stage AD.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/metabolism , Early Growth Response Protein 1/metabolism , Acetylcholinesterase/physiology , Alzheimer Disease/physiopathology , Animals , Brain/pathology , Disease Models, Animal , Disease Progression , Early Growth Response Protein 1/physiology , Frontal Lobe/pathology , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism
6.
Psychoneuroendocrinology ; 95: 34-42, 2018 09.
Article in English | MEDLINE | ID: mdl-29793095

ABSTRACT

BACKGROUND: Alterations in peripheral sex hormones may play an important role in sex differences in terms of stress responses and mood disorders. It is not yet known whether and how stress-related brain systems and brain sex steroid levels fluctuate in relation to changes in peripheral sex hormone levels, or whether the different sexes show different patterns. We aimed to investigate systematically, in male and female rats, the effect of decreased circulating sex hormone levels following gonadectomy on acute and chronic stress responses, manifested as changes in plasma and hypothalamic sex steroids and hypothalamic stress-related molecules. METHOD: Experiment (Exp)-1: Rats (14 males, 14 females) were gonadectomized or sham-operated (intact); Exp-2: gonadectomized and intact rats (28 males, 28 females) were exposed to acute foot shock or no stressor; and Exp-3: gonadectomized and intact rats (32 males, 32 females) were exposed to chronic unpredictable mild stress (CUMS) or no stressor. For all rats, plasma and hypothalamic testosterone (T), estradiol (E2), and the expression of stress-related molecules were determined, including corticotropin-releasing hormone, vasopressin, oxytocin, aromatase, and the receptors for estrogens, androgens, glucocorticoids, and mineralocorticoids. RESULTS: Surprisingly, no significant correlation was observed in terms of plasma sex hormones, brain sex steroids, and hypothalamic stress-related molecule mRNAs (p > 0.113) in intact or gonadectomized, male or female, rats. Male and female rats, either intact or gonadectomized and exposed to acute or chronic stress, showed different patterns of stress-related molecule changes. CONCLUSION: Diminished peripheral sex hormone levels lead to different peripheral and central patterns of change in the stress response systems in male and female rats. This has implications for the choice of models for the study of the different types of mood disorders which also show sex differences.


Subject(s)
Gonadal Steroid Hormones/metabolism , Gonadal Steroid Hormones/physiology , Stress, Physiological/physiology , Animals , Aromatase , Brain/metabolism , Corticotropin-Releasing Hormone , Depression , Depressive Disorder , Estradiol/analysis , Female , Hypothalamus/metabolism , Hypothalamus/physiology , Male , Orchiectomy , Ovariectomy , Oxytocin , Rats , Rats, Sprague-Dawley , Receptors, Steroid/analysis , Sex Characteristics , Sex Factors , Testosterone/analysis , Vasopressins
7.
Neuropsychopharmacology ; 42(10): 2064-2071, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28447621

ABSTRACT

Oxytocin (OXT), synthesized in the hypothalamic paraventricular nucleus (PVN) and then released into different brain areas, may play a crucial role in various behaviors and neuropsychiatric disorders, including depression. Testosterone has been proposed by clinical studies to have the opposite effect of oxytocin in these disorders. We began by studying, in the postmortem hypothalamus of fifteen patients with mood disorders and fifteen matched controls, the expression of OXT in the PVN by means of immunocytochemistry (ICC) and the co-localization of OXT and androgen receptor (AR) by means of double labeling ICC. Subsequently, the regulatory effect of AR on OXT gene expression was studied in vitro. We found a higher expression of PVN OXT in the mood disorder patients than in the control subjects, and observed a clear co-localization of AR in OXT-expressing neurons, both in the cytoplasm and in the nucleus. In addition, a significant decrease in OXT-mRNA levels was observed after pre-incubation of the SK-N-SH cells with testosterone. A further potential androgen-responsive element in the human OXT gene promotor was revealed by electrophoretic mobility shift assays and co-transfections in neuroblastoma cells. Finally, in vitro studies demonstrated that AR mediated the down-regulation of OXT gene expression. These results suggest that the fact that OXT and testosterone appear to have opposite effects in neuropsychiatric disorders might be based upon a direct inhibition of AR on OXT transcription, which may provide a novel target for therapeutic strategies in depression.


Subject(s)
Hypothalamus/metabolism , Mood Disorders/metabolism , Oxytocin/metabolism , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cytoplasm/drug effects , Cytoplasm/metabolism , Cytoplasm/pathology , Gene Expression , Humans , Hypothalamus/pathology , Immunohistochemistry , Mood Disorders/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxytocin/genetics , Promoter Regions, Genetic , RNA, Messenger/metabolism , Testosterone/administration & dosage , Testosterone/metabolism
8.
EBioMedicine ; 18: 311-319, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28377228

ABSTRACT

BACKGROUND: Neurophysiological and behavioral processes regulated by hypocretin (orexin) are severely affected in depression. However, alterations in hypocretin have so far not been studied in the human brain. We explored the hypocretin system changes in the hypothalamus and cortex in depression from male and female subjects. METHODS: We quantified the differences between depression patients and well-matched controls, in terms of hypothalamic hypocretin-1 immunoreactivity (ir) and hypocretin receptors (Hcrtr-receptors)-mRNA in the anterior cingulate cortex (ACC) and dorsolateral prefrontal cortex. In addition, we determined the alterations in the hypocretin system in a frequently used model for depression, the chronic unpredictable mild stress (CUMS) rat. RESULTS: i) Compared to control subjects, the amount of hypocretin-immunoreactivity (ir) was significantly increased in female but not in male depression patients; ii) hypothalamic hypocretin-ir showed a clear diurnal fluctuation, which was absent in depression; iii) male depressive patients who had committed suicide showed significantly increased ACC Hcrt-receptor-2-mRNA expression compared to male controls; and iv) female but not male CUMS rats showed a highly significant positive correlation between the mRNA levels of corticotropin-releasing hormone and prepro-hypocretin in the hypothalamus, and a significantly increased Hcrt-receptor-1-mRNA expression in the frontal cortex compared to female control rats. CONCLUSIONS: The clear sex-related change found in the hypothalamic hypocretin-1-ir in depression should be taken into account in the development of hypocretin-targeted therapeutic strategies.


Subject(s)
Depressive Disorder, Major/pathology , Orexins/metabolism , Aged , Aged, 80 and over , Animals , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Corticosterone/blood , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Depressive Disorder, Major/metabolism , Disease Models, Animal , Female , Gyrus Cinguli/metabolism , Humans , Hypothalamus/metabolism , Immunohistochemistry , Male , Middle Aged , Orexin Receptors/genetics , Orexin Receptors/metabolism , Orexins/genetics , Prefrontal Cortex/metabolism , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sex Characteristics
9.
Behav Brain Res ; 284: 231-7, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25687843

ABSTRACT

Sex differences play an important role in depression, the basis of which is an excessive stress response. We aimed at revealing the neurobiological sex differences in the same study in acute- and chronically-stressed rats. Female Sprague-Dawley (SD) rats were randomly divided into 6 groups: chronic unpredictable mild stress (CUMS), acute foot shock (FS) and controls, animals in all 3 groups were sacrificed in proestrus or diestrus. Male SD rats were randomly divided into 3 groups: CUMS, FS and controls. Comparisons were made of behavioral changes in CUMS and control rats, plasma levels of corticosterone (CORT), testosterone (T) and estradiol (E2), and of the hypothalamic mRNA-expression of stress-related molecules, i.e. estrogen receptor α and ß, androgen receptor, aromatase, mineralocorticoid receptor, glucocorticoid receptor, corticotropin-releasing hormone, arginine vasopressin and oxytocin. CUMS resulted in disordered estrus cycles, more behavioral and hypothalamic stress-related molecules changes and a stronger CORT response in female rats compared with male rats. Female rats also showed decreased E2 and T levels after FS and CUMS, while male FS rats showed increased E2 and male CUMS rats showed decreased T levels. Stress affects the behavioral, endocrine and the molecular response of the stress systems in the hypothalamus of SD rats in a clear sexual dimorphic way, which has parallels in human data on stress and depression.


Subject(s)
Rats, Sprague-Dawley/physiology , Rats, Sprague-Dawley/psychology , Sex Characteristics , Stress, Psychological/physiopathology , Acute Disease , Animals , Body Weight/physiology , Chronic Disease , Corticosterone/blood , Electroshock , Estradiol/blood , Estrous Cycle/physiology , Female , Foot , Hypothalamus/physiopathology , Male , RNA, Messenger/metabolism , Random Allocation , Testosterone/blood
10.
Neurobiol Aging ; 33(11): 2585-98, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22284987

ABSTRACT

Histamine is produced by the hypothalamic tuberomamillary nucleus (TMN). We studied its involvement in Alzheimer's disease (AD) by in situ hybridization of histidine decarboxylase (HDC), the key enzyme of histamine production, in 9 AD patients and 9 controls. Additionally, messenger (m) RNA levels of the 4 histamine receptors (H(1-4)R) and of the enzyme involved in histamine metabolism, histamine methyltransferase (HMT), were determined by quantitative polymerase chain reaction (qPCR) in the prefrontal cortex (PFC) in the course of AD (n = 49). Moreover, alterations in glia markers were studied. HDC-mRNA levels in the TMN were unchanged in AD, despite of the reduced number of Nissl-stained neurons (p = 0.001). However, a decrease in HDC-mRNA was observed in its medial part (mTMN; p = 0.047). In the course of AD only females had increased prefrontal cortex expression of histamine receptor-3 (H(3)R) (p = 0.007) and histamine methyltransferase-mRNA (p = 0.011) and of the glia markers, glial fibrillary acidic protein-mRNA, vimentin-mRNA and proteolipid protein-mRNA. These findings indicate the presence of regional changes in the TMN that are at least partly gender-dependent.


Subject(s)
Alzheimer Disease/metabolism , Histamine/metabolism , Histidine Decarboxylase/metabolism , Receptors, Histamine/metabolism , Aged , Aged, 80 and over , Autopsy , Female , Histamine N-Methyltransferase/metabolism , Histidine Decarboxylase/genetics , Humans , Hypothalamus/metabolism , Male , Middle Aged , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Histamine/genetics
11.
Exp Gerontol ; 46(2-3): 178-84, 2011.
Article in English | MEDLINE | ID: mdl-20833237

ABSTRACT

Our hypothesis is that there is 'wear and tear' in the brain, which is the basis of the process of aging, but that stimulation of brain function may slow down brain aging and diminish the risk for neurodegenerative diseases such as Alzheimer's disease (AD), possibly by activating repair mechanisms. Evidence supporting this hypothesis is presented in this review. During normal aging and in AD, cell loss is not as prominent a phenomenon as is often presumed. In fact, unaltered neuronal numbers have been reported in many brain areas in AD, e.g. in the nucleus basalis of Meynert (NBM) where the number of large neurons decreases while that of small neurons increases. Decreased neuronal activity is an essential characteristic of AD, and a substantial decrease of cerebral glucose metabolism may even precede cognitive impairments. Some hypothalamic neurons remain intact and active during the process of aging, others become even hyperactive, which may lead to disorders. Arginine vasopressin (AVP) levels were found to be higher in the elderly than in young subjects. There is an age-related, sex-dependent activation of the AVP neurons in the supraoptic nucleus (SON) and in the paraventricular nucleus (PVN), which may be the basis of analogous changes in the prevalence of hypertension and hyponatraemia in the elderly. No significant functional loss of magnocellular hypothalamic neurosecretory neurons were found in the SON or PVN in AD. The activity of the corticotrophin releasing hormone (CRH) neurons in the hypothalamic PVN is the basis for the activity of the hypothalamo-pituitary-adrenal (HPA) axis, which is activated during aging in a sex-dependent way, and even more activated in AD. The activated HPA axis is a risk for depression. Environmental stimulation increases brain reserve. An increase in time spent on intellectual activities was associated with a significant decrease in probability to get AD, and occupation has even a stronger indication of diminished risk for dementia. A series of observations showed that a dysfunctional clock may underlie the disordered rhythms in AD. Additional bright light improved the rest-activity rhythms, while giving bright light and/or melatonin to AD patients ameliorated the progression of cognitive and noncognitive symptoms. This implies that neurons affected by AD can still be reactivated if the right stimuli are applied. Unknown diffusible factors from the neural stem cells improve the survival of aged and degenerating neurons in postmortem human brain slice cultures. Gene therapy with nerve growth factor aimed at the NBM showed metabolic activation of various brain regions. A microarray study of the prefrontal cortex in the course of AD revealed an increased expression of genes related to synaptic activity and changes in plasticity during the very early pre-symptomatic stages, which is proposed to represent a coping mechanism against increased soluble ß-amyloid levels. In brief, these examples of the 'use it or lose it' principle during the course of aging or AD now provide novel targets for the development of therapeutic strategies aiming at the prevention and treatment of AD.


Subject(s)
Aging/physiology , Brain/growth & development , Hypothalamus/physiology , Neurons/physiology , Aging/pathology , Brain/pathology , Dementia/physiopathology , Female , Humans , Hypothalamus/growth & development , Male
12.
Vitam Horm ; 82: 339-65, 2010.
Article in English | MEDLINE | ID: mdl-20472147

ABSTRACT

The neuropeptides corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP) are crucially involved in the pathogenesis of depression. The close correlation between the etiology of depression and dysregulation of the stress responses is based upon a hyperactivity of the hypothalamo-pituitary-adrenal (HPA) axis. CRH neurons in the paraventricular nucleus are the motor of the HPA-axis. Centrally released CRH, AVP, and increased levels of cortisol all contribute to the signs and symptoms of depression. Single-nucleotide polymorphisms in the CRH and AVP receptor genes are associated with the risk for depression. Activation of the HPA-axis is generally regarded to be the final common pathway of the pathogenesis of depression. Sex hormones are crucially involved in the regulation of CRH gene expression. The decreased activity of the biological clock, the suprachiasmatic nucleus, as indicated by its lower AVP expression, is the basis for the disturbed rhythms in depression. Both similarities and differences are found in the activity changes in the CRH and AVP systems in depressive disorders and depression in Alzheimer's disease.


Subject(s)
Arginine Vasopressin/metabolism , Corticotropin-Releasing Hormone/metabolism , Depressive Disorder/metabolism , Hypothalamus/metabolism , Alzheimer Disease/complications , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Animals , Depressive Disorder/complications , Depressive Disorder/diagnosis , Depressive Disorder/psychology , Female , Humans , Hypothalamo-Hypophyseal System/metabolism , Male , Pituitary-Adrenal System/metabolism , Postmortem Changes , Sex Characteristics , Sex Factors , Stress, Psychological/diagnosis , Stress, Psychological/metabolism , Stress, Psychological/psychology
13.
Biol Psychiatry ; 66(9): 832-9, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19596122

ABSTRACT

BACKGROUND: Corticotropin-releasing hormone (CRH) is considered the central driving force in the stress response and plays a key role in the pathogenesis of depression. Retinoic acid (RA) has been suggested by clinical studies to be associated with affective disorders. METHODS: First, hypothalamic tissues of 12 patients with affective disorders and 12 matched control subjects were studied by double-label immunofluorescence to analyze the expression of CRH and retinoic acid receptor-alpha (RAR-alpha). Second, critical genes involved in the RA signaling pathways were analyzed in a rat model of depression. Finally, the regulatory effect of RAR-alpha on CRH gene expression was studied in vitro. RESULTS: We found that the expression of RAR-alpha was colocalized with CRH neurons in human hypothalamic paraventricular nucleus (PVN). The density of RAR-alpha-immunoreactive neurons and CRH-RAR-alpha double-staining neurons was significantly increased in the PVN of patients with affective disorders. The ratio of the CRH-RAR-alpha double-staining neurons to the CRH-immunoreactive neurons in affective disorder patients was also increased. Recruitment of RAR-alpha by the CRH promoter was observed in the rat hypothalamus. A dysregulated RA metabolism and signaling was also found in the hypothalamus of a rat model for depression. Finally, in vitro studies demonstrated that RAR-alpha mediated an upregulation of CRH gene expression. CONCLUSIONS: These results suggest that RAR-alpha might contribute to regulating the activity of CRH neurons in vivo, and the vulnerable character of the critical proteins in RA signaling pathways might provide novel targets for therapeutic strategies for depression.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Mood Disorders/metabolism , Mood Disorders/physiopathology , Receptors, Retinoic Acid/metabolism , Up-Regulation , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase 1 Family , Animals , Arginine Vasopressin/metabolism , Cell Line, Tumor , Female , Humans , Hypothalamus/metabolism , Male , Middle Aged , Mood Disorders/genetics , Neurons/metabolism , Rats , Receptors, Retinoic Acid/physiology , Retinal Dehydrogenase/metabolism , Retinoic Acid Receptor alpha , Signal Transduction/genetics , Stress, Physiological/physiology , Tretinoin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL